CN108727584B - Anti-cancer conjugates - Google Patents

Anti-cancer conjugates Download PDF

Info

Publication number
CN108727584B
CN108727584B CN201710263126.1A CN201710263126A CN108727584B CN 108727584 B CN108727584 B CN 108727584B CN 201710263126 A CN201710263126 A CN 201710263126A CN 108727584 B CN108727584 B CN 108727584B
Authority
CN
China
Prior art keywords
cancer
tumor
pharmaceutically acceptable
drug conjugate
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201710263126.1A
Other languages
Chinese (zh)
Other versions
CN108727584A (en
Inventor
袁建栋
黄仰青
宋云松
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Borui Biomedical Suzhou Co ltd
Original Assignee
Borui Biomedical Suzhou Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to CN201710263126.1A priority Critical patent/CN108727584B/en
Application filed by Borui Biomedical Suzhou Co ltd filed Critical Borui Biomedical Suzhou Co ltd
Priority to PCT/CN2018/083746 priority patent/WO2018192550A1/en
Priority to JP2020504758A priority patent/JP6947909B2/en
Priority to ES18787379T priority patent/ES2859473T3/en
Priority to EP18787379.9A priority patent/EP3613792B1/en
Priority to US16/498,765 priority patent/US11191843B2/en
Priority to KR1020197032879A priority patent/KR102279429B1/en
Priority to AU2018255458A priority patent/AU2018255458B2/en
Priority to CA3058029A priority patent/CA3058029A1/en
Publication of CN108727584A publication Critical patent/CN108727584A/en
Application granted granted Critical
Publication of CN108727584B publication Critical patent/CN108727584B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G65/00Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule
    • C08G65/02Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring
    • C08G65/32Polymers modified by chemical after-treatment
    • C08G65/329Polymers modified by chemical after-treatment with organic compounds
    • C08G65/333Polymers modified by chemical after-treatment with organic compounds containing nitrogen
    • C08G65/33303Polymers modified by chemical after-treatment with organic compounds containing nitrogen containing amino group
    • C08G65/33317Polymers modified by chemical after-treatment with organic compounds containing nitrogen containing amino group heterocyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G65/00Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule
    • C08G65/02Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring
    • C08G65/32Polymers modified by chemical after-treatment
    • C08G65/329Polymers modified by chemical after-treatment with organic compounds
    • C08G65/333Polymers modified by chemical after-treatment with organic compounds containing nitrogen
    • C08G65/3332Polymers modified by chemical after-treatment with organic compounds containing nitrogen containing carboxamide group
    • C08G65/33327Polymers modified by chemical after-treatment with organic compounds containing nitrogen containing carboxamide group cyclic
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G65/00Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule
    • C08G65/02Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring
    • C08G65/32Polymers modified by chemical after-treatment
    • C08G65/329Polymers modified by chemical after-treatment with organic compounds
    • C08G65/333Polymers modified by chemical after-treatment with organic compounds containing nitrogen
    • C08G65/33396Polymers modified by chemical after-treatment with organic compounds containing nitrogen having oxygen in addition to nitrogen

Abstract

The invention discloses a structure withA multi-branched drug conjugate of formula (i) or a pharmaceutically acceptable salt thereof:r is an organic center, POLY is a polymer, L is a multivalent linker, T is a targeting molecule, D is an active agent, q is any integer between 3 and 8, wherein L is:

Description

Anti-cancer conjugates
Technical Field
The invention relates to a multi-arm polymer modified targeting anticancer conjugate, in particular to a conjugate formed by connecting a targeting molecule and an anticancer drug through a multi-arm polymer.
Background
Over the years, various methods have been proposed for improving the stability and delivery of bioactive agents. Challenges associated with the formulation and delivery of pharmaceutical agents may include: poor water solubility, toxicity, low bioavailability, instability, and rapid in vivo degradation of the pharmaceutical agent. Although many approaches have been devised to improve the delivery of pharmaceutical agents, none of the individual approaches have had their drawbacks. For example, drug delivery methods commonly employed aim to solve or at least ameliorate one or more of the following problems, including drug encapsulation, such as in a liposome, polymer matrix, or unimolecular micelle, covalent attachment to a water soluble polymer such as polyethylene glycol, use of gene targeting agents, salt structures, and the like.
WO2005028539, WO2010019233, WO2011063156, WO2011063158 disclose a drug nktr 102 in the third clinical stage, which is mainly used for metastatic breast cancer and developed by Nektar Therapeutics. The drug is a water-soluble multi-branched polymer prodrug to improve the load of the drug, and has the following structure:
Figure BDA0001275323580000011
the compound is connected with irinotecan by multi-arm PEG, so as to improve water solubility, increase drug loading and reduce side effects under the condition of unchanged anticancer effect. However, the drug still has the disadvantages of poor targeting, incapability of acting on specific cancer cells, capability of killing the cancer cells and influencing the performance of normal cells, and high incidence of adverse reaction.
Disclosure of Invention
The invention discloses a brand-new targeting multi-branched-chain drug conjugate, which has three or more branched chains and can be expressed as the following formula:
Figure BDA0001275323580000021
r is an organic center, POLY is a polymer, L is a multivalent linker, T is a targeting molecule, D is an active agent, q is any integer between 3 and 8, wherein L is:
Figure BDA0001275323580000022
the symbol "#" represents the point of attachment of the multivalent linker L to the targeting molecule T, "#" represents the point of attachment of the multivalent linker L to the active agent D, "%" represents the point of attachment of the multivalent linker L to POLY, wherein L is any integer between 2 and 20, and m and n are each any integer between 0 and 10;
d is camptothecin drug shown as formula (II):
Figure BDA0001275323580000023
R1-R5independently of one another, from the following groups: hydrogen, halogen, acyl, alkyl, substituted alkyl, alkoxy, substituted alkoxy, alkenyl, alkynyl, cycloalkyl, hydroxy, cyanoNitro, azido, amide, hydrazine, amine, substituted amine, hydroxycarbonyl, alkoxycarbonyl, alkoxycarbonyloxy, carbamoyloxy, arylsulfonyloxy, alkylsulfonyloxy; r6Is H OR OR8,R8Is alkyl, alkenyl, cycloalkyl, haloalkyl or hydroxyalkyl; r7Is hydroxyl, amino, or thiol.
POLY is a polymer, L is a multivalent linker, T is a targeting molecule, D is an active agent, and the four together form the "branches" of the multi-branched drug conjugate. Each of the branches and the other branches of the multi-branched drug conjugate are independent of each other. Each branch emanates from an organic center "R". However, in general, each branch of the conjugate is the same.
Each of the variable moieties in structural formula (I) will now be described in detail.
Organic center, "R"
In formula (I), "R" is an organic core group of 1 to 100 atoms. Preferably, R contains from 3 to 50 atoms, and more preferably, R contains from about 3 to 30 atoms. R may be a core of all carbon atoms, and may optionally contain 1 or more heteroatoms, e.g., O, S, N, P, etc., depending on the particular central molecule used. R may be linear, branched or cyclic, giving rise to at least 3 independent polymer branches. In formula (I), "q" corresponds to the number of polymer branches emanating from "R".
The organic center "R" is derived from a molecule that provides a number of polymer attachment sites, approximately equal to the number of polymer branches. More preferably, the main central formula of the multi-chain polymer structure is at least the residue of a polyol, polysulfide or polyamine compound having 3 and more than 3 hydroxyl, thio or amino groups suitable as polymer branches. A "polyol" is a molecule consisting of a plurality (greater than 2) of available hydroxyl groups. A "polysulfide" is a molecule that consists of a plurality (greater than 2) of available thio groups. A "polyamine compound" is a molecule consisting of a plurality (greater than 2) of available amine groups. Depending on the number of polymeric branches, the precursor of the polyol, polyamine compound or polysulfide (prior to covalent bonding of POLY) typically comprises from 3 to 25 hydroxyl, thio or amino groups, preferably from 3 to 10 hydroxyl, thio or amino groups, and most preferably from 3 to about 8 (e.g., 3, 4, 5, 6, 7 or 8) hydroxyl, thio or amino groups suitable for covalent bonding with POLY.
The precursor of the polyol or polyamine centre typically has a formula R- (OH) p or R- (NH) prior to reaction with the polymer2) p is the same as the formula (I). In formula (I), the values of p and q correspond, since each functional group in the parent organic molecule typically has an-OH and-NH group2If the positions are susceptible or susceptible to reaction, they are covalently bonded to POLY of the polymer branches. In formula (I), the hydroxyl groups of the polyol of the R parent have been converted to a polymer branch after attachment to POLY, and R is depicted as the residue after attachment. For example, if the organic center molecule is derived from pentaerythritol, the precursor of the polyol has the formula C (CH)2OH)4The organic central group R is represented by:
Figure BDA0001275323580000041
illustrative preferred polyhydroxy compounds as the polymer center include aliphatic polyhydroxy compounds containing 1 to 10 carbon atoms and 1 to 10 hydroxyl groups, for example, ethylene glycol, alkylene glycols, hydrocarbon glycols, alkylene hydrocarbon glycols, hydrocarbon cycloalkyl glycols, 1, 5-decalin glycols, 4, 8-bis (hydroxymethyl) tricyclodecane, cycloalkylene glycols, dihydroxyalkanes, trihydroxyalkanes, tetrahydroxyalkanes, and the like. Cycloaliphatic polyols include straight or closed ring sugars and sugar alcohols such as mannitol, sorbitol, cellosolve, xylitol, bromitol, threitol, arabitol, erythritol, hexitol, ribose, arabinose, xylose, lyxose, rhamnose, galactose, glucose, fructose, sorbose, mannose, pyranose, altrose, talose, tagatose, pyranoside, sucrose, lactose, maltose and the like. Aromatic polyols such as phenylphosphonium diol, hydrocarbyl phenylphosphonium diol, pyrogallol, fluoroglycinol, 1,2, 4-benzenetriol, resorcinol, hydrocarbyl resorcinol, dihydrocarbyl resorcinol, orcinol monohydrate, olivetol, hydroquinone, hydrocarbyl hydroquinone, phenyl hydroquinone, and the like may also be used. Other polyol centers that may be employed may include crown ethers, cyclodextrins, dextrins, or other carbohydrates.
In the structural formula (I), q is the number of polymer branches connected to corresponding R, and the specific number can be 3-20. Typically, specific numbers for "q" are 3, 4, 5, 6, 7, 8. Specifically, three, four, five, six, seven, eight polymer branches are emitted centering on "R".
In some embodiments, "R" has three polymer branches, "R" is preferably:
Figure BDA0001275323580000042
in some embodiments, "R" has four polymer branches, "R" is preferably:
Figure BDA0001275323580000043
in some embodiments, "R" has six polymer branches, "R" is preferably:
Figure BDA0001275323580000051
in some embodiments, "R" has eight polymer branches, "R" is preferably:
Figure BDA0001275323580000052
polymer, "POLY"
In formula (I), "POLY" is a polymer and the POLY in each polymer branch is independently selected, preferably each polymer is the same polymer, more preferably each polymer branch in formula (I) is the same. Preferred polymers are water soluble, any water soluble polymer may be used to form the conjugates of the invention, and any polymer referred to herein may be of any geometry or form. Representative polymers include, but are not limited to: polyethylene glycol, polypropylene glycol, poly (vinylpyrrolidone), poly (hydroxyalkyl methacrylate amine), poly (hydroxyalkyl methacrylate), poly (saccharide), poly (± -hydroxy acid), poly (acrylic acid), poly (vinyl acetic acid), polyphosphazine, polyoxazoline, poly (N-acryloylmorpholine), and the like.
In typical compounds, "POLY" is polyethylene glycol (PEG) and can be in any geometric form or form, including straight, branched, forked, etc., and "polyethylene glycol" as used herein is meant to encompass any water-soluble POLY (ethylene oxide). Typically, the PEG used in the present invention will comprise one of two of the following structures: "(CH)2CH2O)k- "or" (CH)2CH2O)k-CH2CH2- "depending on whether one or more of the terminal oxygens has been replaced, for example, during a synthetic transformation. The variable k ranges from 5 to about 500, and the terminal groups as well as the structure of the overall PEG can vary. The polyethylene glycol structure will typically also contain a partial terminal moiety residue, similar to the terminal group of POLY, which may be H, NH2、OH、CO2H、C1-6Alkyl (e.g. methyl, ethyl, propyl), C1-6Alkoxy (e.g., methoxy, ethoxy), acyl, or aryl.
Preferred "POLY" of the present invention are linear polyethylene glycols, typically of the structure:
Figure BDA0001275323580000053
represents a link of an atomJoint, mark "&The oxygen atom of the number "is the atom attached to the organic center" R ". Wherein k is in the range of about 5 to 500, preferably 50 to 200, r is any integer between 1 and 10, and more preferably, the term "POLY" according to the present invention is:
Figure BDA0001275323580000061
POLY of the present invention may also be:
Figure BDA0001275323580000062
Figure BDA0001275323580000063
and the like.
The active agent D is a camptothecin anticancer agent, and camptothecin drugs are clinical topoisomerase I inhibitors, have the defects of high activity, poor water solubility, high toxic and side effects on normal body tissues and the like, and greatly limit the clinical application of the camptothecin anticancer agent.
R in the structure of D7Is a group covalently attached to a multivalent linker L, such as a hydroxyl, amino, or thiol, preferably a hydroxyl group. There should be no significant loss of biological activity when active agent D is attached to the multivalent linker L.
The active agent of the invention is preferably irinotecan, SN-38, 10-hydroxycamptothecin and rubitecan.
Wherein the structure of irinotecan is as follows:
Figure BDA0001275323580000064
the structure of SN-38 is as follows:
Figure BDA0001275323580000065
the structure of 10-hydroxycamptothecin is as follows:
Figure BDA0001275323580000071
rubitecan has the following structure:
Figure BDA0001275323580000072
in the invention, the T is a targeting molecule with or without medicinal effect, and the targeting molecule has the effect of increasing targeting property, so that the concentration of the conjugate in a target tissue is higher, and the physiological activity or the medicinal effect is improved. The "T" may be a monofunctional targeting molecule or a multifunctional targeting molecule, and in some alternatives, may also be a targeting moiety consisting of two or more targeting molecules. For example, T may be GE 11.
The polypeptide sequence of GE11 is: YHWYGYTPQNVI, its structure is:
Figure BDA0001275323580000073
the active agent "D" referred to herein refers to a portion of the unmodified parent active agent or the residue of the unmodified parent active agent prior to the covalent chain (or activated or chemically modified form thereof) resulting from covalent attachment of the drug to the multivalent linker of the present invention. The active agent itself is released upon hydrolysis or enzymatic hydrolysis of the linker between the active agent moiety and the multivalent linker.
For the purposes of the present invention, the term "residue" is understood to mean a part of a compound which is the residue after having undergone a substitution reaction with another compound.
When the conjugate of the present invention enters into an organism and reaches a target cell or a target tissue, the active agent D is cleaved from the polyvalent linker L, and the active agent D is released in a form in which it is not modified, i.e., a covalent bond is not formed, is separated from the parent body, and exerts physiological activity.
In a preferred embodiment of the invention, "POLY" is a linear polyethylene glycol linker arm, i.e., the conjugates of the invention include several types of compounds:
four arms:
Figure BDA0001275323580000081
three arms:
Figure BDA0001275323580000082
eight arms:
Figure BDA0001275323580000091
wherein k is in the range of about 5 to 500, preferably 50 to 200, and r is any integer between 1 and 10.
The compounds of the formula (III) are preferred according to the invention, and k is preferably 113 on the basis of the formula (III). It will be understood by those skilled in the art that in the polymer art, k represents the degree of polymerization of the polymer and is not an absolute number depending on the molecular weight of the polymer, and when k is 113, it means an average value of 113.
In a more preferred embodiment, the targeting moiety "T" of the conjugate of the invention is GE11 and the active agent "D" is selected from one of irinotecan, SN-38, 10-hydroxycamptothecin, rubitecan.
In a more preferred embodiment, L is selected from:
Figure BDA0001275323580000092
Figure BDA0001275323580000101
one kind of (1).
Based on formula (iii), in some particular embodiments, the compounds of the invention are as follows:
a compound f: d is irinotecan, T is GE11
Figure BDA0001275323580000102
Compound F is a pharmaceutically acceptable hydrochloride salt of compound F
Figure BDA0001275323580000103
More specifically, compound f can also be written as follows:
Figure BDA0001275323580000111
it is to be noted here that, in the salt formation, the side chain of the conjugate of the invention and HCl are respectively salified, for example, compound F, each side chain carries 3 molecules of HCl, and the whole molecule will carry 12 HCl molecules.
In addition to the specific compounds disclosed above, those skilled in the art can prepare more conjugates according to the technical scheme and preparation method of the present invention, such as:
d is SN-38, T is GE11 conjugate;
② D is 10-hydroxycamptothecin, T is GE11 conjugate;
③ D is rubitecan and T is GE11 conjugate.
The conjugate of the invention is a typical prodrug, and the active agent D is released through hydrolysis or enzymolysis, separated from a parent body and exerts physiological activity.
The conjugates of the invention exhibit high loading capacity, which allows for a reduction in the total dose to treat a particular disease, such as cancer, etc. That is, the active agent carriers of the conjugates of the invention are capable of effectively covalently bonding to a wide variety of active agent molecules, allowing a greater number of therapeutic dosage forms (i.e., active agent moieties) to be administered per a given amount of conjugate. The conjugate is modified by the water-soluble polymer, is hydrophilic in nature, and particularly improves the bioavailability of the conjugate when the active agent is a water-insoluble drug.
Compared with unconjugated drugs, the conjugate of the invention can show stronger effect and is more enriched in tissues in human bodies or other animal bodies.
The conjugate prodrugs of the present invention contain a number of unique properties, particularly where the active agent is an anti-cancer compound. The drug precursor can inhibit the growth of tumor with high efficiency. This small molecule we use is one known to have anti-cancer properties. However, by combining with multi-branched polymers as described above, the therapeutic efficacy and pharmacokinetics are greatly improved compared to the small molecule (e.g., the anti-cancer compound itself). Suitable solid tumor types include colon cancer, breast cancer, ovarian cancer, pancreatic cancer, gastric cancer, brain glioma, and malignant sarcomas, carcinomas, and lymphomas of the breast, ovary, colon, kidney, bile duct, lung, and brain.
In conclusion, the invention is a multi-arm polymer modified targeting anticancer conjugate, wherein the water-solubility polymer modification can enhance the water solubility of the conjugate, thereby improving the drug loading rate; the targeting molecule increases targeting property, so that the concentration of the conjugate in target tissues is higher; l is any connecting joint and has the function of connecting the targeting molecule and the anticancer drug firstly and then connecting the targeting molecule, the anticancer drug and the polymer arm, so that the whole conjugate forms an organic whole.
The conjugate of the invention, the pharmaceutically acceptable salt is preferably hydrochloride, can be salified by conventional means in the field of pharmaceutical chemistry, and can also be trifluoroacetate, sulfate, phosphate, acetate and the like.
In another aspect, the invention provides a method for preparing the conjugate. During the preparation of the conjugates of the invention, POLY and organic center R actually comprise a multi-armed polymer, which in the preferred embodiment of the invention is a multi-armed polyethylene glycol, and can be obtained from commercially available starting materials, such as various types of four-, three-, eight-armed polyethylene glycol derivatives available from Kyork technology, Inc., Beijing. These multi-arm PEGs, which are commercially available, can participate directly in the reaction.
In preparing the conjugate of formula (III), the four-armed polyethylene glycol preferably used is as follows:
Figure BDA0001275323580000131
the preferred four-ARM polyethylene glycol is referred to as 4ARM-PEG20K-SCM and has a molecular weight of about 20 kDa. Similarly, in the preparation of the conjugates of formula (IV) and formula (V), the three-and eight-arm polyethylene glycols used also preferably have a molecular weight of about 20 kDa.
DETAILED DESCRIPTION OF EMBODIMENT (S) OF INVENTION
The present invention will be described in detail below. This invention may, however, be embodied in many different forms and should not be construed as limited to the embodiments set forth herein, which are provided for the purpose of making the disclosure more complete and complete. The reagents and raw materials are commercially available except for the preparation method, wherein 4ARM-PEG20K-SCM is available from Kyork technology Co., Ltd
Noun interpretation
DMF: n, N-dimethylformamide
DCM: methylene dichloride
Boc-Gly-OH:
Figure BDA0001275323580000141
DMAP: 4-dimethylaminopyridine
DCC: dicyclohexylcarbodiimide
IPA: isopropanol (I-propanol)
TFA: trifluoroacetic acid
TBME: tert-butyl methyl ether
EA: ethyl acetate
DME: ethylene glycol dimethyl ether
Fmoc-OSU: 9-fluorenylmethyl-N-succinimidyl carbonate
THF: tetrahydrofuran (THF)
H-Lys(Boc)-OBzl·HCl:
Figure BDA0001275323580000142
DIEA: n, N-diisopropylethylamine
DEPC: cyanophosphoric acid diethyl ester
DEA: triethylamine
Pbf:
Figure BDA0001275323580000143
HOBT: 1-hydroxybenzotriazoles
DIC: n, N-diisopropylcarbodiimide
TFE: trifluoroethanol
DPPA: azoic acid diphenyl ester
SPPS: solid phase organic synthesis
NMM: n-methylmorpholine
And (3) TIS: tri-isopropyl silane
MTBE: tert-butyl methyl ether
Example 1
Figure BDA0001275323580000151
Preparation of Compound 2
Adding 3.50g of compound 1(1.0eq) and 52.5mL of DMF into a 250mL round-bottom flask, heating to 60 ℃ for dissolution, distilling off the DMF under reduced pressure after 5-10min, adding 300mL of n-heptane, distilling under reduced pressure for three times, adding 105mL of DCM, 1.08g of Boc-Gly-OH (1.2eq) and 63mg of DMAP (0.1eq) after spin-drying, adding 1.59g of DCC (1.5eq) dissolved in 10mL of DCM, reacting at 20 ℃ for 4 hours, filtering after TLC monitoring reaction, adding 120mL of IPA when the reaction is finished, distilling off 75% of the solvent, adding 150mL of n-heptane, stirring at room temperature for 1 hour, filtering, washing with n-heptane for 2 times, and drying to obtain 4.02g of compound 2 as a light yellow solid.
Preparation of Compound 3
Adding 4.02g of compound 2 and 50mL of DCM into a 100mL three-necked flask, stirring and dissolving, then adding 11.6mL of TFA dropwise, reacting for 2h at room temperature, monitoring by TLC, adding 150mL of acetonitrile after the reaction is finished, distilling 120mL of solvent under reduced pressure, pouring into 320mL of TBME solution, stirring for 30min, filtering, and washing a filter cake with TBME to obtain 4.00g of light yellow solid compound 3.
Example 2
Figure BDA0001275323580000161
Preparation of Compound 5
Adding 6.9g of compound 4 and 30mL of EA into a 250mL three-necked flask, stirring to dissolve, cooling to 0 ℃, adding 40mL of 0.3M HCl/EA, keeping the temperature for reaction for 2 hours, and concentrating to dryness after the TLC monitoring reaction to obtain a compound 5, and directly carrying out the next reaction.
Preparation of Compound 6
Dissolving compound 5(1.0eq) with 50ml of purified water, adding 3.96g of sodium bicarbonate (2.0eq), dissolving 5.30g of Fmoc-OSU (1.0eq) with 50ml of DME, adding into a reaction flask of compound 5, supplementing with 25ml of THF, stirring at room temperature for 2 hours, after TLC monitoring reaction is finished, evaporating organic solvent, EA extracting impurities, adjusting pH of an aqueous phase to 3-4 with dilute hydrochloric acid, EA extracting for 2 times, combining organic phases, washing with water once, washing with saturated saline, drying with anhydrous sodium sulfate, and concentrating to obtain 8.4g of light yellow oily compound 6.
Preparation of Compound 7
In a 100ml reaction flask were charged 4.00g of Compound 6(1.0eq), 2.92g H-Lys (Boc) -OBzl & HCl, and 40ml of DCM to dissolve them, and 2.76g of DIEA (3.0eq), 1.74g of DEPC (1.5eq) were added thereto, and the mixture was stirred at room temperature for 2 hours, after the TLC-monitored reaction was completed, it was washed with an aqueous acetic acid solution, with a sodium hydrogencarbonate solution, once with water, once with a saturated saline solution, and then dried over anhydrous sodium sulfate, and concentrated to obtain 7.0g of Compound 7 as a pale yellow oil, which was then subjected to the next reaction without being purified. (Compound 16 was prepared by the same method)
Preparation of Compound 8
After completion of TLC monitoring reaction, the mixture was concentrated to dryness, 100ml of 50ml of EA was added thereto, pH was adjusted to 3-4 with dilute hydrochloric acid, liquid separation was performed, and the aqueous phase was extracted with EA for 2 times and concentrated to dryness to obtain 3.5g of Compound 8 as a pale yellow solid. (Compound 17 was prepared by the same method)
Example 3
Preparation of protecting group-attached targeting molecule GE11 (Compound 60)
Figure BDA0001275323580000171
Sequence YHWYGYTPQNVI of GE11
The following protected amino acids were attached to the Resin in sequence using 2Cl-Trt Resin, HOBT/DIC as coupling reagent, DMF as reaction solvent, ninhydrin assay for reaction monitoring: Fmoc-Ile-OH, Fmoc-Val-OH, Fmoc-Asn (Trt) -OH, Fmoc-Gln (Trt) -OH, Fmoc-Pro-OH, Fmoc-Thr (tBu) -OH, Fmoc-Tyr (tBu) -OH, Fmoc-Gly-OH, Fmoc-Tyr (tBu) -OH, Fmoc-Trp (Boc) -OH, Fmoc-His (Trt) -OH, Boc-Tyr (tBu) -OH, addition of a cleavage reagent: acetic acid/TFE/DCM ═ 1/2/7, reaction for 2 hours, precipitation with ice MTBE, washing, and drying to give off-white solid 60.
EXAMPLE 4 preparation of Compound F and Compound F
Figure BDA0001275323580000181
Figure BDA0001275323580000191
Preparation of Compound 61
5.00g of compound 60(1.0eq), 1.66g of compound 17(1.1eq), 50mL of DMF, 0.72g of DIEA (3.0eq), 0.45g of DEPC (1.5eq) are added into a 100mL reaction flask, and reacted at room temperature for 2 hours, after the TLC reaction is finished, the mixture is poured into 10mL of water, EA is extracted for 2 times, acetic acid aqueous solution is washed, sodium bicarbonate solution is washed, saturated sodium chloride is washed, anhydrous sodium sulfate is dried, and jelly-like solid 616.14 g is obtained by concentration, and the next reaction is directly carried out.
Preparation of Compound 62
Adding 6.10g of compound 61, 150ml of methanol and 0.31g of Pd/C into a 200ml hydrogenation kettle, hydrogenating overnight, filtering after TLC reaction is finished, and concentrating to obtain 625.98 g of gray solid compound
Preparation of Compound 63
5.95g of compound 62(1.0eq), 1.36g of compound 3(1.05eq), 60ml of DMF, 0.68g of DIEA (3.0eq), 0.43g of DEPC (1.5eq) are added into a 100ml reaction flask, the mixture is reacted for 2 hours at room temperature, the mixture is poured into 600ml of TBME after TLC reaction is finished, the mixture is pumped and filtered after being pulped, the solid is dissolved by 100ml of DCM and poured into 1.0L of TBME, the mixture is pumped and filtered after being pulped, 636.53 g of gray powder compound is dried, and the next reaction is directly carried out.
Preparation of Compound 64
6.50g of Compound 63 130ml of lysis reagent: acetic acid/TFE/DCM ═ 1/2/7, reaction 2 hours, precipitation with ice MTBE, washing, drying and HPLC purification gave 3.63g of off-white solid compound 64. Preparation of Compound 65
2.06g of compound 64(4.5eq), 3.00g of 4ARM-PEG20K-SCM (1.0eq), 30ml of DMF (dimethyl formamide), 0.13g of TEA (9.0eq) are added into a 50ml reaction bottle to react at room temperature, after the reaction is monitored by HPLC to have no obvious progress, the mixture is poured into 300ml of TBME, pulped, filtered by suction and dried to obtain 654.67 g of compound, and the compound is directly used for the next reaction.
Synthesis of Compound f
Into a 200ml reaction flask, 4.60g of compound 65, 100ml lysis reagent: 92.5% TFA/2.5% water/2.5% TIS, stirred at room temperature for 2h, precipitated with ice MTBE, centrifuged, washed, crude product purified by reverse phase HPLC, desalted, concentrated to remove organic solvent, and lyophilized to give 1.34g of off-white powder compound f.
Synthesis of Compound F
And (3) purifying the crude compound F by reverse phase HPLC, desalting, concentrating to remove the organic solvent, adjusting the pH value to 5-6 by using dilute hydrochloric acid, and freeze-drying to obtain 1.34g of a yellow-green powder compound F.
MALDI-TOF molecular weight 30907.82.
EXAMPLE 5 evaluation of the in vivo efficacy of Compounds in HT-29 nude mouse graft tumor model
1. Purpose of experiment
Evaluation of efficacy of Compound F and Compound F following administration in mouse animal models of xenografted BALB/c nude of HT-29 cell line of human Colon cancer.
2. Experimental Material
2.1 test article
Irinotecan (bulk drug) was purchased and nktr-102 and 2 compounds tested were provided by borrelid biopharmaceutical (suzhou) corporation.
The preparation method of nktr-102 refers to the method disclosed in CN102711837A, and comprises the following steps:
compound 3(829mg,4.5eq) from example 1 was added to a 250mL reaction flask, DCM (50mL), triethylamine (221mg,9.0eq) were added, and after dissolution 4ARM-PEG20K-SCM (5.00g,1.0eq) was added to the reaction flask. After no significant progress of the reaction was monitored by HPLC, about 20mL of DCM was distilled off under reduced pressure, the solution was poured into 300mL of TBME and precipitated with stirring, filtered to give 5.4g of crude product, which was purified by HPLC preparative, desalted, adjusted to pH 5-6 with dilute hydrochloric acid, and lyophilized to give 2.71g of light green powder nktr-102.
2.2 reagents
McCoy's 5A culture solution, Fetal Bovine Serum (FBS), trypsin, cyan-chain double antibody, water for injection, normal saline, lactic acid and sorbitol.
2.3 Experimental animals
Female BALB/c nude mice (the number is 150; the week age is 6-7 weeks) are purchased from Beijing Wintolite laboratory animal technology Limited, and are bred in an SPF animal room, the temperature is 20-25 ℃, the relative humidity is 40-70%, and the illumination is carried out for 12 hours respectively; animals had free access to water and food. After about 1 week of normal feeding, mice with good signs can be selected for this experiment by veterinary examination. Marking the tail and root of the animals by using a marker pen before grouping, and marking each animal after grouping by using an ear clipping mode.
2.4 transplantable tumor strains
Human colon cancer cells HT-29, from the cell bank of the Committee for culture Collection of the Chinese academy of sciences (CAS, the laboratory liquid Nitrogen cryopreservation).
3. Experimental methods
3.1HT-29 cell culture
At 5% CO2HT-29 cells are subjected to conventional cell culture in a culture solution containing 10% fetal bovine serum McCoy's 5A under the culture condition of 37 ℃; passage with 0.25% pancreatin; according to the growth condition of the cells, the cells are passaged 2 to 3 times per week at the passage ratio of 1:4 to 1: 6.
3.2 animal model preparation
Collecting HT-29 cells in logarithmic growth phase, counting the cells, suspending in serum-free McCoy's 5A culture medium, and adjusting the cell concentration to 4 × 107cell/mL; blowing and beating the cells by using a pipettor to disperse the cells uniformly, then putting the cells into a 50mL centrifuge tube, and putting the centrifuge tube into an ice box; the cell suspension was aspirated by a 1mL syringe, injected subcutaneously into the right forelimb armpit of nude mice, and inoculated with 100. mu.L (4X 10) per animal6Cell/cell), establishing a HT-29 nude mouse transplantation tumor model. After inoculation, the animal state and the tumor growth condition are regularly observed, the tumor diameter is measured by using an electronic vernier caliper, the data is directly input into an Excel spreadsheet, and the tumor volume is calculated. When the tumor volume reaches 100-300 mm330 animals with good health status and similar tumor volume were selected and divided into 5 groups (n is 6) by the randomized block method. Tumor size was measured 2 times per week after the start of the experiment, tumor volume was calculated, and animal body weight was weighed and recorded.
Tumor Volume (TV) calculation formula is as follows:
TV(mm3)=l×w2/2
wherein l represents the tumor major axis (mm); w represents the tumor minor diameter (mm).
3.3 preparation of solvent
0.5g of sorbitol was weighed into a 50mL centrifuge tube, 50mL of water for injection was added to the centrifuge tube, and the solid matter was dissolved completely by vortex oscillation to prepare a 1% sorbitol aqueous solution (w/v) and stored in a refrigerator at 4 ℃ for future use.
3.4 preparation of drug delivery preparation
3, 4.1 irinotecan administration preparation
12.0mg of irinotecan was weighed, 0.15mL of 1% lactic acid was added, the mixture was vortexed to completely dissolve the drug, 2.85mL of 1% sorbitol aqueous solution was added, and the mixture was vortexed and mixed uniformly, whereby the ratio of 1% lactic acid to 1% sorbitol aqueous solution in the solution was about 5:95 (v/v). The effective concentration of irinotecan in the solution is 4.0 mg/mL-1
3.4.2nktr-102 administration preparation
Before each administration, 101.5mg of nktr-102 was accurately weighed, 2.5mL of physiological saline was added, and the solution was vortexed to completely dissolve the drug, whereby the effective concentration of irinotecan in the solution was 4.0 mg. multidot.mL-1
3.4.3 formulation of administration of Compound a and Compound A
Before each administration, respectively accurately weighing 120.3mg of compound a and compound A, adding 2.5mL of physiological saline, vortex vibrating, and ultrasonically (if necessary) completely dissolving the medicine, wherein the effective concentration of irinotecan in the solution is 4.0 mg/mL-1
3.4.4 formulation of Compound F and Compound F for administration
Before each administration, 145.9mg of compound F is accurately weighed, 2.5mL of physiological saline is added, vortex oscillation and ultrasonic treatment (if necessary) are carried out to completely dissolve the medicine, and the effective concentration of irinotecan in the solution is 4.0 mg/mL-1
3.5 animal grouping and administration
Animal groups and dosing regimens are shown in table 1. The first administration is started on the grouping day, the experiment is finished after about 21 days, and the administration volumes are 10 mL/kg-1. The effective dose of the reduced irinotecan is 40mg kg-1. The first group was a solvent control group, and the saline was administered to the tail vein 1 time every 4 days for 3 times (Q4D × 3). In groups 2 to 5, test samples of irinotecan, nktr-102, compound F, compound F were administered by tail vein injection, each once every 4 days, Q4D X3.
TABLE 1 drug effect experiment dosing regimen for nude mouse transplanted tumor model
Figure BDA0001275323580000231
3.6 end of experiment
On the last day of the experiment, the animals were weighed, and euthanized after tumor size measurement (CO)2). And stripping and weighing tumor tissues, and calculating the tumor weight inhibition rate.
4. Data recording, calculation formula
The formula for the Relative Tumor Volume (RTV) is:
RTV=TVt/TVinitial
wherein, TVinitialTumor volume measured when administered in groups; TV (television)tThe tumor volume at each measurement during dosing.
The relative tumor proliferation rate (% T/C) was calculated by the following formula:
%T/C=100%×(RTVT/RTVC)
wherein, RTVTRepresenting treatment group RTV; RTVCRepresenting the solvent control RTV.
The calculation formula of the tumor growth inhibition rate TGI (%) is as follows:
TGI=100%×[1-(TVt(T)-TVinitial(T))/(TVt(C)-TVinitial(C))]
wherein, TVt(T)Represents the tumor volume for each measurement in the treatment group; TV (television)initial(T)Represents the tumor volume of the treatment group when administered in groups; TV (television)t(C)Represents the tumor volume of each measurement of the solvent control group; TV (television)initial(C)The tumor volume of the solvent control group at the time of group administration is indicated.
The formula for calculating the weight loss rate of the animals is as follows:
weight loss rate of animal (BW) 100 × (BW)initial-BWt)/BWinitial
Wherein, BWtRepresents the animal body weight measured at each time during the dosing period; BW (Bandwidth)initialThe body weight of the animals at the time of the group administration is indicated.
The calculation formula of the tumor weight inhibition rate IR (%) is as follows:
IR(%)=100%×(WC-WT)/WC
wherein, WCRepresenting tumor weight of control group; wTIndicates the tumor weight of the treated group.
5. Statistical analysis method
Experimental data were calculated and statistically processed using Microsoft Office Excel 2007 software. Data are expressed as Mean ± standard error (Mean ± SE) and comparisons between groups are performed using the t-test, unless otherwise specified.
6. Experimental observation
During the course of the experiment, the laboratory and veterinarians need to continuously observe the signs and health of the experimental animals. Any abnormal manifestations of the animal, such as pain, depression, decreased activity, etc., need to be recorded in the original record of the experiment. If the abnormal performance of the experimental animal exceeds the welfare of the IACUC-related animal, the veterinarian can determine whether to terminate the experiment and notify the responsible person of the experimental project.
7. Results
For the human cancer xenograft tumor model, the relative tumor proliferation rate T/C (%) is recommended as the test evaluation index, and the lower the proliferation rate, the better the tumor inhibition effect, see table 2.
TABLE 2 tumor proliferation rate T/C (%)
Figure BDA0001275323580000241
Figure BDA0001275323580000251
P <0.05 compared to RTV of the group of blank solvent, irinotecan and nktr-102
# P <0.05 compared to% T/C for the blank solvent, irinotecan, and nktr-102 group
Experimental results show that the compound has good inhibition effect on the in-vivo growth of human colon cancer HT-29 nude mouse transplantation tumor model tumor, and is superior to irinotecan and nktr-102.
Example 6 inhibition of human Breast cancer MDA-MB-231 nude mouse xenograft model
1. Purpose of experiment
This study evaluated the in vivo anti-tumor activity of compound F and compound F using a human breast cancer MDA-MB-231 nude mouse graft tumor model.
2. Laboratory animal
2.1 animal species
A mouse.
2.2 variety
BALB/c nude mice.
2.3 sex
And (4) female.
2.4 number
150 were inoculated and 30 were used for the experiment.
Age 2.5
6-8 weeks.
2.6 body weight
20-22 g. + -. 20% body weight mean.
2.7 animal sources (suppliers)
Shanghai Sphere-BiKai laboratory animals Co., Ltd (BK), license number SCXK (Shanghai) 2008-0016.
2.8 management of laboratory animals
All experimental animals were housed in an SPF scale laboratory. The experimenters were responsible for routine care and experimental studies.
2.8.1 animal identity identification method
Each mouse cage is hung with an identity card with information such as experiment number, experiment group, name of experimenter, mouse variety and sex, and the mouse is marked with an ear nail.
2.8.2 random grouping
When the tumor volume reaches 150-3Then dividing the mice into 5 groups by a random block method, wherein each group comprises 6 mice, and ensuring that the tumor volume and the weight of the mice among the groups are uniform. The mean tumor volumes of the groups differed from the mean tumor volumes of all experimental animals by no more than ± 10%.
2.8.3 operation management Specification
All experimental animals strictly adhere to the guidelines for animal use and management.
2.8.4 feeding conditions
The living conditions are as follows: IVC system, 6 per cage
Temperature: 20-26 deg.C
Humidity: 40% +/-70%
Illumination: 12 hours of day and night alternation
2.8.5 feed
Irradiated rat and rat feeds were purchased from Aojieli feeds, Inc., Beijing, Ke. Free food intake.
2.8.6 Drinking water
City tap water is filtered, autoclaved and sterilized for drinking.
2.8.7 bedding
Corncobs, Shanghai Miao derivatives science and technology Co., Ltd, were used after autoclaving. The pads were changed twice a week.
2.8.8 acclimation period
Mice were given a minimum one week environmental acclimation period prior to the experiment.
3. Experimental Material
3.1 test drugs
Irinotecan (bulk drug) was purchased and nktr-102 and 2 compounds tested were provided by borrelid biopharmaceutical (suzhou) corporation.
3.2 other chemical reagents and materials
3.2.1 physiological saline
Physiological saline was purchased from Shanghai Huayuan Changfu pharmaceutical industry (group) Co., Ltd.
3.2.2 sterile Syringe
A1 ml sterile syringe was purchased from Shanghai Kangdelai Enterprise development group, Inc. (Shanghai, China).
3.2.3 cell lines
Human breast cancer MDA-MB-231 was purchased from the Shanghai institute of cell biology.
MDA-MB-231 was cultured in DMEM medium (GIBCO, USA) containing 10% fetal bovine serum FBS (GIBCO, USA) and in 5% CO237 ℃ incubator.
3.2.4 Matrigel (BD Matrigel)
Matrigel was purchased from BD corporation, usa
3.3 instruments
Biosafety cabinets (model: AC2-6E1), available from ESCO;
CO2a water-tight cell culture chamber (model: 3111) from Thermo Scientific Forma;
an inverted microscope (model: CKX41SF) from Olympus;
electric suction apparatus (model YX930D), available from Shanghai medical devices industry (group) Inc.;
a balance (Mettler-Torloduo AB135-S) available from Mettler-Torluo;
low speed centrifuge (model LD5-2A) available from Beijing Rebo centrifuge, Inc.;
an electronic digital display caliper (model: SF2000) purchased from Guilin Guangdong digital measurement and control GmbH.
4. Design of experiments
Establishing a model of human breast cancer MDA-MB-231 nude mice subcutaneous transplantation tumor, wherein each nude mouse is inoculated with 1 × 106And (4) cells.
The following (table 3) doses and dosing schedules were designed for this trial.
Table 3: anti-tumor effect of human breast cancer MDA-MB-231 in nude mouse transplantation tumor model
Figure BDA0001275323580000271
5. Compound preparation
The formulation method is provided by borry biomedical technology (Suzhou) Inc.
3mL volume required for a single administration.
5.1 formulation of irinotecan administration preparation
12.0mg of irinotecan was weighed, 0.15mL of 1% lactic acid was added, the mixture was vortexed to completely dissolve the drug, 2.85mL of 1% sorbitol aqueous solution was added, and the mixture was vortexed and mixed uniformly, whereby the ratio of 1% lactic acid to 1% sorbitol aqueous solution in the solution was about 5:95 (v/v). The concentration of irinotecan in the free form in the solution was 4.0 mg/mL-1
5.2nktr-102 dosing formulations
Before each administration, 101.5mg of nktr-102 was accurately weighed, 2.5mL of physiological saline was added, and the solution was vortexed to completely dissolve the drug, and the concentration of irinotecan in the solution was 4.0 mg. multidot.mL in the free form-1
5.3 preparation of compound F and compound F for administration: accurately weighing, adding 2.5mL of physiological saline, and vortex vibrating to completely dissolve the medicine, wherein the free form concentration of irinotecan in the solution is 4.0 mg/mL-1
6. Experimental methods
MDA-MB-231 cells were cultured in DMEM containing 10% fetal bovine serum FBS (GIBCO, USA). Cells were placed in 5% CO2The culture was carried out in an incubator at 37 ℃.
Establishing a tumor nude mouse subcutaneous transplantation model by a cell inoculation method: collecting tumor cells in logarithmic growth phase, counting, re-suspending in 1 × PBS, and adjusting cell suspension concentration to 1 × 107And/ml. The right dorsal part of the nude mice was inoculated subcutaneously with 1X 10 tumor cells using a 1ml syringe (No. 4 needle)60.1 ml/mouse.
The tumor volume reaches 100-200mm3When the animals are divided into 5 groups according to a random block method, the tumor difference of each group is less than 10 percent of the mean value, 6 animals in each group are divided into 6 animals, the current Day is used as Day1, and the animals are administrated on the current Day.
The experimental period was carried out for 3 weeks, during which animal body weight and tumor size were measured twice a week. Clinical symptoms were recorded with daily observations. Animals were sacrificed the last day of the experiment, weighed, tumors stripped, weighed and photographed for documentation.
All animal experimental procedures strictly adhere to animal use and management regulations. The calculation of the tumor related parameters refers to the CFDA technical guidance principle of non-clinical research of cytotoxic antitumor drugs in China.
The Tumor Volume (TV) is calculated as: TV as a × b2/2. Where a, b represent tumor measurement length and width, respectively. The Relative Tumor Volume (RTV) is calculated as: RTV-Vt/V0. Wherein V0Tumor volume at the time of group administration and Vt is the tumor volume at the time of measurement. Having antitumor activityThe evaluation indexes are relative tumor increment rate T/C (%) and tumor inhibition rate (%), and the calculation formulas are respectively as follows: T/C (%) ═ TRTV/CRTV)×100%。TRTVTo the treatment group RTV, CRTVNegative control group RTV; tumor inhibition (%) (average tumor weight in negative control group-average tumor weight in administration group)/average tumor weight in negative control group × 100%.
The change (%) in body weight of tumor-bearing animals was calculated as follows: (weight-at-measurement-weight-at-group)/weight-at-group × 100.
7. Data analysis
Experimental data were calculated and statistically processed using Microsoft Office Excel 2007 software. Data are expressed as Mean ± standard error (Mean ± SE) and comparisons between groups are performed using the t-test, unless otherwise specified.
8. Results and reports
According to the Chinese CFDA non-clinical research technical guide principle of cytotoxic antitumor drugs (11 months 2006), T/C (percent) is less than or equal to 40 percent, and p is less than 0.05 through statistical analysis to be effective, which is shown in Table 4.
TABLE 4 tumor proliferation rate T/C (%)
Figure BDA0001275323580000291
P <0.05 compared to RTV of the group of blank solvent, irinotecan and nktr-102
# P <0.05 compared to% T/C for the blank solvent, irinotecan, and nktr-102 group
Experimental results show that the compound has good inhibition effect on human breast cancer MDA-MB-231 nude mouse transplanted tumor, and is superior to irinotecan and nktr-102.
Example 7 inhibition of human pancreatic cancer MIA Paca-2 nude mouse xenograft model
1. Purpose of experiment
This study evaluated the in vivo anti-tumor activity of compound F and compound F using a human pancreatic cancer MIA Paca-2 nude mouse transplantation tumor model.
2. Laboratory animal
2.1 animal species
A mouse.
2.2 variety
BALB/c-nu/nu nude mice.
2.3 sex
And (4) female.
2.4 number
150。
2.5. Age (age)
6-8 weeks.
2.6. Body weight
20-22 g. + -. 20% body weight mean.
2.7. Animal origin (supplier)
Shanghai Sphere-BiKai laboratory animals Co., Ltd (BK), license number SCXK (Shanghai) 2008-0016.
2.8. Laboratory animal management
All experimental animals were housed in an SPF scale laboratory.
2.8.1 animal identity identification method
Each mouse cage is hung with an identity card with information such as experiment number, experiment group, name of experimenter, mouse variety and sex, and the mouse is marked with an ear nail.
2.8.2 random grouping
When the tumor volume reaches 150-3Then dividing the mice into 5 groups by a random block method, wherein each group comprises 6 mice, and ensuring that the tumor volume and the weight of the mice among the groups are uniform. The mean tumor volumes of the groups differed from the mean tumor volumes of all experimental animals by no more than ± 10%.
2.8.3 operation management Specification
The operation and management of all experimental animals strictly comply with the guiding principles of the use and management of the experimental animals.
2.8.4 feeding conditions
The living conditions are as follows: IVC system, 6 per cage
Temperature: 25 ℃ plus or minus 1 DEG C
Humidity: 65% +/-10%
Illumination: 12 hours of day and night alternation
2.8.5 feed
Irradiated rat and rat feeds were purchased from Aojieli feeds, Inc., Beijing, Ke. Free food intake.
2.8.6 Drinking water
City tap water is filtered, autoclaved and sterilized for drinking.
2.8.7 bedding
Corncobs, Shanghai Miao derivatives science and technology Co., Ltd, were used after autoclaving. The pads were changed twice a week.
2.8.8 acclimation period
Mice were given a minimum one week environmental acclimation period prior to the experiment.
3. Experimental Material
3.1 test drugs
Irinotecan (bulk drug) was purchased and nktr-102 and 2 compounds tested were provided by borrelid biopharmaceutical (suzhou) corporation.
3.2 other chemical reagents and materials
3.2.1 physiological saline
Physiological saline was purchased from Shanghai Huayuan Changfu pharmaceutical industries (group) Co., Ltd. (Shanghai, China).
3.2.2 sterile Syringe
A1 ml sterile syringe was purchased from Shanghai Kangdelai Enterprise development group, Inc. (Shanghai, China).
3.2.3 cell lines
Human pancreatic cancer MIA Paca-2 was purchased from Shanghai institute of sciences cell biology.
MIA Paca-2 was cultured in DMEM medium (GIBCO, USA) containing 10% fetal bovine serum FBS (GIBCO, USA) and 2.5% HS in 5% CO237 ℃ incubator.
3.2.4 Matrigel (BD Matrigel)
Matrigel was purchased from BD corporation, usa
3.3 instruments
Biosafety cabinets (model: AC2-6E1), available from ESCO;
CO2a water-tight cell culture chamber (model: 3111) from Thermo Scientific Forma;
an inverted microscope (model: CKX41SF) from Olympus;
electric suction apparatus (model YX930D), available from Shanghai medical devices industry (group) Inc.;
a balance (Mettler-Torloduo AB135-S) available from Mettler-Torluo;
low speed centrifuge (model LD5-2A) available from Beijing Rebo centrifuge, Inc.;
an electronic digital display caliper (model: SF2000) purchased from Guilin Guangdong digital measurement and control GmbH.
4. Design of experiments
Establishing a human pancreatic cancer MIA Paca-2 nude mouse subcutaneous transplantation tumor model, and inoculating 3X 10 tumor cells to each nude mouse6And (4) cells.
The following (table 5) doses and dosing schedules were designed for this trial.
Table 5: antitumor effect in human pancreatic cancer MIA Paca-2 nude mouse transplantation tumor model
Tumor strain: MIA Paca-2; a total of 30 inoculations
Figure BDA0001275323580000321
5. Compound preparation
The formulation method is provided by borry biomedical technology (Suzhou) Inc.
A single administration requires a volume of 3 mL.
5.1 formulation of irinotecan administration preparation
12.0mg of irinotecan was weighed, 0.15mL of 1% lactic acid was added, the mixture was vortexed to completely dissolve the drug, 2.85mL of 1% sorbitol aqueous solution was added, and the mixture was vortexed and mixed uniformly, whereby the ratio of 1% lactic acid to 1% sorbitol aqueous solution in the solution was about 5:95 (v/v). The concentration of irinotecan in the free form in the solution was 4.0 mg/mL-1
5.2nktr-102 dosing formulations
Before each administration, 101.5mg of nktr-102 was accurately weighed, 2.5mL of physiological saline was added, and the solution was vortexed to completely dissolve the drug, and the concentration of irinotecan in the solution was 4.0 mg. multidot.mL-1 in the free form.
5.3 Compounds f andf, preparation of a dosing preparation: accurately weighing, adding 2.5mL of physiological saline, and vortex vibrating to completely dissolve the medicine, wherein the free form concentration of irinotecan in the solution is 4.0 mg/mL-1.6. Experimental methods
MIA Paca-2 cells were cultured in DMEM containing 10% fetal bovine serum FBS (GIBCO, USA) and 2.5% HS. Cells were placed in 5% CO2The culture was carried out in an incubator at 37 ℃.
Establishing a tumor nude mouse subcutaneous transplantation model by a cell inoculation method: collecting tumor cells in logarithmic growth phase, counting, re-suspending in 1 × PBS, and adjusting cell suspension concentration to 3 × 107And/ml. Nude mice were inoculated subcutaneously on the right dorsal side with tumor cells, 3X 10, using a 1ml syringe (No. 4 needle)60.1 ml/mouse.
The tumor volume reaches 100-200mm3When the animals are divided into 5 groups according to a random block method, the tumor difference of each group is less than 10 percent of the mean value, 6 animals in each group are divided into 6 animals, the current Day is used as Day1, and the animals are administrated on the current Day.
The experimental period was carried out for 3 weeks, during which animal body weight and tumor size were measured twice a week. Clinical symptoms were recorded with daily observations. Animals were sacrificed the last day of the experiment, weighed, tumors stripped, weighed and photographed for documentation.
All animal experimental procedures strictly adhere to animal use and management regulations. The calculation of the tumor related parameters refers to the SFDA non-clinical research technical guidance principle of cytotoxic antitumor drugs in China.
The Tumor Volume (TV) is calculated as: TV as a × b2/2. Where a, b represent tumor measurement length and width, respectively. The Relative Tumor Volume (RTV) is calculated as: RTV-Vt/V0. Wherein V0Tumor volume at the time of group administration and Vt is the tumor volume at the time of measurement. The evaluation indexes of the antitumor activity are relative tumor increment rate T/C (%) and tumor inhibition rate (%), and the calculation formulas are respectively as follows: T/C (%) ═ TRTV/CRTV)×100%。TRTVTo the treatment group RTV, CRTVNegative control group RTV; tumor inhibition ratio (%) (average tumor weight in negative control group-average tumor weight in administration group)/average tumor weight in negative control group100%。
The change (%) in body weight of tumor-bearing animals was calculated as follows: (weight-at-measurement-weight-at-group)/weight-at-group × 100.
According to the Chinese SFDA non-clinical research technical guide principle of cytotoxic antitumor drugs (11 months 2006), T/C (percent) is less than or equal to 40 percent, and P <0.05 is analyzed by statistics to be effective.
7. Data analysis
Experimental data were calculated and statistically processed using Microsoft Office Excel 2007 software. Data are expressed as Mean ± standard error (Mean ± SE) unless otherwise stated, and comparisons between groups were by t-test with P <0.05 as a significant difference.
8. Results and reports
According to the Chinese CFDA non-clinical research technical guide principle of cytotoxic antitumor drugs (11 months 2006), T/C (percent) is less than or equal to 40 percent, and P <0.05 is analyzed to be effective by statistics, which is shown in Table 6.
TABLE 6 tumor proliferation rate T/C (%)
Figure BDA0001275323580000331
Figure BDA0001275323580000341
P <0.05 compared to RTV of the group of blank solvent, irinotecan and nktr-102
# P <0.05 compared to% T/C for the blank solvent, irinotecan, and nktr-102 group
Experimental results show that the compound has good inhibition effect on human pancreatic cancer MIA Paca-2 nude mouse transplantation tumor, and is superior to irinotecan and nktr-102.
Example 8 inhibition of tumor growth in a nude mouse transplanted tumor model of human gastric cancer NCI-N87 cell line.
1. Purpose of experiment
The inhibitory effect of the test drugs compound F and compound F on the in vivo growth of the tumor of the nude mouse transplanted tumor model of the human gastric cancer NCI-N87 cell line is evaluated.
2. Experimental Material
2.1 test article
Irinotecan (bulk drug) was purchased and, thus, nktr-102 and 2 compounds tested were provided by borrelid biopharmaceutical (suzhou) corporation.
2.2 reagents
RPMI-1640 culture medium, Fetal Bovine Serum (FBS), trypsin, cyan-chain double antibody and normal saline.
2.3 Experimental animals
Female BALB/c nude mice (the number is 150; the week age is 6-8 weeks) are purchased from Beijing Wintolite laboratory animal technology, Inc., and are bred in an SPF animal house of St.Su.Sco, Suzhou, the temperature is 20-25 ℃, the relative humidity is 40-70%, and the illumination is carried out for 12 hours respectively; animals had free access to water and food. After about 1 week of normal feeding, mice with good signs can be selected for this experiment by veterinary examination. Marking the tail and root of the animals by using a marker pen before grouping, and marking each animal after grouping by using an ear clipping mode.
2.4 transplantable tumor strains
Human gastric cancer cell NCI-N87, derived from the cell bank of the Committee for culture Collection of the Chinese academy of sciences (CAS, the laboratory is frozen in liquid nitrogen).
3 Experimental methods
3.1 NCI-N87 cell culture
At 5% CO2Under the culture condition of 37 ℃, NCI-N87 cells are cultured in a culture solution containing 10 percent fetal calf serum RPMI-1640 by a conventional method; passage with 0.25% pancreatin; according to the growth condition of the cells, the cells are passaged 1 to 2 times per week at the passage ratio of 1:2 to 1: 6.
3.2 animal model preparation
Collecting NCI-N87 cells in logarithmic growth phase, counting the cells, suspending in serum-free RPMI-1640 medium, and adjusting the cell concentration to 5 × 107cell/mL; blowing and beating the cells by using a pipettor to disperse the cells uniformly, then putting the cells into a 50mL centrifuge tube, and putting the centrifuge tube into an ice box; the cell suspension was aspirated by a 1mL syringe and injected subcutaneously into the right forelimb axilla of nude mice, one animalInoculation with 100. mu.L (5X 10)6Cell/cell), establish NCI-N87 nude mouse graft tumor model. After inoculation, the animal state and the tumor growth condition are regularly observed, the tumor diameter is measured by using an electronic vernier caliper, the data is directly input into an Excel spreadsheet, and the tumor volume is calculated. When the tumor volume reaches 100-300 mm330 animals with good health status and similar tumor volume were selected and divided into 5 groups (n is 6) by the randomized block method. Tumor size was measured 2 times per week after the start of the experiment, tumor volume was calculated, and animal body weight was weighed and recorded.
Tumor Volume (TV) calculation formula is as follows:
TV(mm3)=l×w2/2
wherein l represents the tumor major axis (mm); w represents the tumor minor diameter (mm).
3.3 preparation of solvent
0.5g of sorbitol was weighed into a 50mL centrifuge tube, 50mL of water for injection was added to the centrifuge tube, and the solid matter was dissolved completely by vortex oscillation to prepare a 1% sorbitol aqueous solution (w/v) and stored in a refrigerator at 4 ℃ for future use.
3.4 preparation of drug delivery preparation
3, 4.1 irinotecan administration preparation
12.0mg of irinotecan was weighed, 0.15mL of 1% lactic acid was added, the mixture was vortexed to completely dissolve the drug, 2.85mL of 1% sorbitol aqueous solution was added, and the mixture was vortexed and mixed uniformly, whereby the ratio of 1% lactic acid to 1% sorbitol aqueous solution in the solution was about 5:95 (v/v). The concentration of irinotecan in the free form in the solution was 4.0 mg/mL-1
3.4.2nktr-102 administration preparation
Before each administration, 101.5mg of nktr-102 was accurately weighed, 2.3mL of physiological saline was added, and the solution was vortexed to completely dissolve the drug, and the concentration of irinotecan in the solution was 4.0 mg. multidot.mL in the free form-1
3.4.3 preparation of compound F, compound F for administration: accurately weighing, adding 2.5mL of physiological saline, and vortex vibrating to completely dissolve the irinotecan in the solution at a free form concentration of 4.0 mg/mL-1
3.5 animal grouping and administration
Animal groups and dosing regimens are shown in table 7. The first administration was started on the day of grouping, and the experiment was ended after 21 days, and the administration volumes were all 10 mL/kg-1. Group 1 was a solvent control group, and the solvent was administered to the tail vein by injection 1 time every 4 days for 3 times (Q4D × 3). In groups 2 to 5, test samples of irinotecan, nktr-102 and the test compound were administered by tail vein injection at a dose of 40 mg/kg-1(calculated as irinotecan content), Q4D × 3.
TABLE 7 drug effect experiment dosing regimen for nude mouse transplanted tumor model
Figure BDA0001275323580000361
3.6 end of experiment
After the experiment was completed, the animals were weighed, and the animals were euthanized after tumor size measurement (CO)2). And stripping and weighing tumor tissues, and calculating the tumor weight inhibition rate. Tumor tissues were weighed and transferred to a sub-70 ℃ freezer for storage for subsequent analysis.
4. Data recording, calculation formula
The formula for the Relative Tumor Volume (RTV) is:
RTV=TVt/TVinitial
wherein, TVinitialTumor volume measured when administered in groups; TV (television)tThe tumor volume at each measurement during dosing.
The relative tumor proliferation rate (% T/C) was calculated by the following formula:
%T/C=100%×(RTVT/RTVC)
wherein, RTVTRepresenting treatment group RTV; RTVCRepresenting the solvent control RTV.
The calculation formula of the tumor growth inhibition rate TGI (%) is as follows:
TGI=100%×[1-(TVt(T)-TVinitial(T))/(TVt(C)-TVinitial(C))]
wherein, TVt(T)Represents the tumor volume for each measurement in the treatment group; TV (television)initial(T)Represents the tumor volume of the treatment group when administered in groups; TV (television)t(C)Represents the tumor volume of each measurement of the solvent control group; TV (television)initial(C)The tumor volume of the solvent control group at the time of group administration is indicated.
The formula for calculating the weight loss rate of the animals is as follows:
weight loss rate of animal (BW) 100 × (BW)initial-BWt)/BWinitial
Wherein, BWtRepresents the animal body weight measured at each time during the dosing period; BW (Bandwidth)initialThe body weight of the animals at the time of the group administration is indicated.
The calculation formula of the tumor weight inhibition rate IR (%) is as follows:
IR(%)=100%×(WC-WT)/WC
wherein, WCRepresenting tumor weight of control group; wTIndicates the tumor weight of the treated group.
5. Statistical analysis method
Experimental data were calculated and statistically processed using Microsoft Office Excel 2007 software. Data are expressed as Mean ± standard error (Mean ± SE) and the two comparisons are performed using t-test, unless otherwise specified.
6. Experimental observation
During the course of the experiment, the laboratory and veterinarians need to continuously observe the signs and health of the experimental animals. Any abnormal manifestations of the animal, such as pain, depression, decreased activity, etc., need to be recorded in the original record of the experiment. If the abnormal performance of the experimental animal exceeds the welfare of the IACUC-related animal, the veterinarian can determine whether to terminate the experiment and notify the responsible person of the experimental project.
7. Results
For the human cancer xenograft tumor model, the relative tumor proliferation rate T/C (%) is recommended as the test evaluation index, and the lower the proliferation rate, the better the tumor inhibition effect, see table 8.
TABLE 8 tumor proliferation rate T/C (%)
Figure BDA0001275323580000371
Figure BDA0001275323580000381
P <0.05 compared to RTV of the group of blank solvent, irinotecan and nktr-102
# P <0.05 compared to% T/C for the blank solvent, irinotecan, and nktr-102 group
Experimental results show that the compound has good inhibition effect on tumor growth of a nude mouse transplantation tumor model of human gastric cancer NCI-N87 cell strain, and is superior to irinotecan and nktr-102.
Example 9 effect on survival of U87MG nude mouse brain in situ model.
1. Purpose of experiment
The effect of test agents compound F and compound F on the survival rate of U87MG nude mouse brain in situ model was evaluated.
2. Experimental Material
2.1 test article
Irinotecan (bulk drug) was purchased and, thus, nktr-102 and 2 compounds tested were provided by borrelid biopharmaceutical (suzhou) corporation.
2.2 reagents
RPMI-1640 culture medium, trypsin, cyan-catenin and normal saline.
2.3 Experimental animals
Female BALB/c nude mice (the number is 150; the week age is 6-8 weeks) are purchased from Beijing Wintolite laboratory animal technology Limited, and are bred in an SPF animal room, the temperature is 20-25 ℃, the relative humidity is 40-70%, and the illumination is carried out for 12 hours respectively; animals had free access to water and food. After about 1 week of normal feeding, mice with good signs can be selected for this experiment by veterinary examination. Marking the tail and root of the animals by using a marker pen before grouping, and marking each animal after grouping by using an ear clipping mode.
2.4 transplantable tumor strains
Glioma cells U87MG, derived from the cell bank of the culture Collection of the type (CAS, the laboratory liquid nitrogen frozen stock) of Chinese academy of sciences.
3. Experimental methods
NCI-N87 cell culture
At 5% CO2Under the culture condition of 37 ℃, NCI-N87 cells are cultured in RPMI-1640 culture solution in a conventional way; passage with 0.25% pancreatin; according to the growth condition of the cells, the cells are passaged 1 to 2 times per week at the passage ratio of 1:2 to 1: 6.
3.1 animal model preparation
Collecting NCI-N87 cells in logarithmic growth phase, counting the cells, suspending in serum-free RPMI-1640 medium, and adjusting the cell concentration to 1 × 108cell/mL; blowing and beating the cells by using a pipettor to disperse the cells uniformly, then putting the cells into a 50mL centrifuge tube, and putting the centrifuge tube into an ice box; the cell suspension is sucked by a 1mL syringe, and 1 mu L (1X 10) of the human brain glioma cell U87MG cells cultured in vitro is guided by an animal stereotaxic apparatus by a microinjection method5Cell/cell), a U87MG brain glioma in situ model was established, and the animal status was observed periodically after inoculation. On day 12 post inoculation, 30 animals were selected and divided into 5 groups (n-6) using the randomized block method.
3.2 preparation of drug delivery preparation
3.2.1 formulation of irinotecan administration preparation
12.0mg of irinotecan was weighed, 0.15mL of 1% lactic acid was added, the mixture was vortexed to completely dissolve the drug, 2.85mL of 1% sorbitol aqueous solution was added, and the mixture was vortexed and mixed uniformly, whereby the ratio of 1% lactic acid to 1% sorbitol aqueous solution in the solution was about 5:95 (v/v). The concentration of irinotecan in the free form in the solution was 4.0 mg/mL-1
3.2.2nktr-102 dosage formulations
Before each administration, 101.5mg of nktr-102 was accurately weighed, 2.5mL of physiological saline was added, and the solution was vortexed to completely dissolve the drug, and the concentration of irinotecan in the solution was 4.0 mg. multidot.mL in the free form-1
3.2.3 preparation of administration of Compound F, Compound F: accurately weighing, adding 2.5mL of physiological saline, and vortexingThe drug is completely dissolved by rotary oscillation, and the free form concentration of irinotecan in the solution is 4.0 mg/mL-1
3.3 animal grouping and administration
Animal groups and dosing regimens are shown in table 9. The first administration was started on the day of grouping, and the experiment was ended after 21 days, and the administration volumes were all 10 mL/kg-1. Group 1 was a solvent control group, and the solvent was administered to the tail vein by injection 1 time every 4 days for 3 times (Q4D × 3). In groups 2 to 5, test samples of irinotecan, nktr-102 and the test compound were administered by tail vein injection at a dose of 40 mg/kg-1(calculated as irinotecan content), Q4D × 3.
TABLE 9 drug effect experiment dosing regimen for nude mouse transplantable tumor model
Figure BDA0001275323580000391
Figure BDA0001275323580000401
4. Data recording, calculation formula
Animal survival time was recorded.
5. Statistical analysis method
Experimental data were calculated and statistically processed using Microsoft Office Excel 2007 software. The two groups were compared using the t-test.
6. Results
See Table 10
TABLE 10 animal survival time (days)
Figure BDA0001275323580000402
P <0.05 median survival time compared to the blank solvent, irinotecan and nktr-102 group
The experimental result shows that the compound has good inhibition effect on brain glioma and is superior to irinotecan and nktr-102.

Claims (14)

1. A multi-branched drug conjugate having the following structural formula (i):
Figure FDA0002778975270000011
r is an organic center, POLY is a polymer, L is a multivalent linker, T is a targeting molecule, D is an active agent, q is any integer between 3 and 8, wherein L is:
Figure FDA0002778975270000012
the symbol "#" represents the point of attachment of the multivalent linker L to the targeting molecule T, "#" represents the point of attachment of the multivalent linker L to the active agent D, "%" represents the point of attachment of the multivalent linker L to POLY, wherein L is any integer between 2 and 20, and m and n are each any integer between 0 and 10;
t is GE 11;
d is camptothecin drug shown as formula (II):
Figure FDA0002778975270000013
R1-R5independently of one another, from the following groups: hydrogen, halogen, acyl, alkyl, substituted alkyl, alkoxy, substituted alkoxy, alkenyl, alkynyl, cycloalkyl, hydroxy, cyano, nitro, azido, amido, hydrazine, amino, substituted amino, hydroxycarbonyl, alkoxycarbonyl, alkoxycarbonyloxy, carbamoyloxy, arylsulfonyloxy, alkylsulfonyloxy; r6Is H OR OR8,R8Is alkyl, alkenyl, cycloalkyl, haloalkyl or hydroxyalkyl; r7Is a hydroxyl group;
"R" is selected from:
Figure FDA0002778975270000014
Figure FDA0002778975270000021
POLY is selected from
Figure FDA0002778975270000022
Figure FDA0002778975270000023
Wherein the content of the first and second substances,
Figure FDA0002778975270000024
representing the junction of atoms, marks "&The oxygen atom of the number is an atom connected to the organic center "R", k is an integer in the range of 50 to 200, and R is any integer between 1 and 10.
2. The multi-branched drug conjugate of claim 1, which is represented by structural formula (iii), (iv), or (v):
Figure FDA0002778975270000025
Figure FDA0002778975270000031
3. the multi-branched drug conjugate of claim 1, wherein POLY is
Figure FDA0002778975270000032
4. The multi-branched drug conjugate of claim 2, or a pharmaceutically acceptable salt thereof, which is:
Figure FDA0002778975270000041
5. the multi-branched drug conjugate according to claim 4, wherein k has an average value of 113, or a pharmaceutically acceptable salt thereof.
6. The multi-branched drug conjugate of claim 5, or a pharmaceutically acceptable salt thereof, wherein L is:
Figure FDA0002778975270000042
7. the multi-branched drug conjugate according to claim 6, wherein D is irinotecan, SN-38, 10-hydroxycamptothecin, or rubitecan, or a pharmaceutically acceptable salt thereof.
8. The multi-branched drug conjugate of claim 7, or a pharmaceutically acceptable salt thereof, which is:
Figure FDA0002778975270000051
and salts thereof.
9. The multi-branched drug conjugate of claim 8, or a pharmaceutically acceptable salt thereof, which is:
Figure FDA0002778975270000052
10. a pharmaceutically acceptable composition comprising the multi-branched drug conjugate of any one of claims 1 to 9, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
11. Use of the multi-branched drug conjugate according to any one of claims 1 to 9 or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of cancer.
12. Use of a multi-branched drug conjugate according to any one of claims 1 to 9, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of colon cancer, lung cancer, breast cancer, ovarian cancer, pancreatic cancer, gastric cancer, brain glioma and malignant sarcomas, carcinomas and lymphomas of the kidney, bile duct and brain.
13. Use of a composition according to claim 10 for the preparation of a medicament for the treatment of cancer.
14. Use of a composition according to claim 10 for the preparation of a medicament for the treatment of colon cancer, lung cancer, breast cancer, ovarian cancer, pancreatic cancer, gastric cancer, brain glioma and malignant sarcomas, carcinomas and lymphomas of the kidney, bile duct and brain.
CN201710263126.1A 2017-04-21 2017-04-21 Anti-cancer conjugates Active CN108727584B (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
CN201710263126.1A CN108727584B (en) 2017-04-21 2017-04-21 Anti-cancer conjugates
JP2020504758A JP6947909B2 (en) 2017-04-21 2018-04-19 Multi-arm targeted anti-cancer conjugate
ES18787379T ES2859473T3 (en) 2017-04-21 2018-04-19 Multi-arm Targeted Anticancer Conjugate
EP18787379.9A EP3613792B1 (en) 2017-04-21 2018-04-19 Multi-arm targeted anti-cancer conjugate
PCT/CN2018/083746 WO2018192550A1 (en) 2017-04-21 2018-04-19 Multi-arm targeted anti-cancer conjugate
US16/498,765 US11191843B2 (en) 2017-04-21 2018-04-19 Multi-arm targeting anti-cancer conjugate
KR1020197032879A KR102279429B1 (en) 2017-04-21 2018-04-19 Multi-cancer target anti-cancer conjugate
AU2018255458A AU2018255458B2 (en) 2017-04-21 2018-04-19 Multi-arm targeted anti-cancer conjugate
CA3058029A CA3058029A1 (en) 2017-04-21 2018-04-19 Multi-arm targeted anti-cancer conjugate

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201710263126.1A CN108727584B (en) 2017-04-21 2017-04-21 Anti-cancer conjugates

Publications (2)

Publication Number Publication Date
CN108727584A CN108727584A (en) 2018-11-02
CN108727584B true CN108727584B (en) 2021-01-05

Family

ID=63933277

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201710263126.1A Active CN108727584B (en) 2017-04-21 2017-04-21 Anti-cancer conjugates

Country Status (1)

Country Link
CN (1) CN108727584B (en)

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1852740B (en) * 2003-09-17 2011-05-11 耐科塔医药公司 Multi-arm polymer prodrugs
WO2007092646A2 (en) * 2006-02-09 2007-08-16 Enzon Pharmaceuticals, Inc. Multi-arm polymeric conjugates of 7-ethyl-10-hydroxycamptothecin for treatment of breast, colorectal, pancreatic, ovarian and lung cancers
KR20090108082A (en) * 2007-02-09 2009-10-14 엔존 파마슈티컬즈, 인코포레이티드 Treatment of resistant or refractory cancers with multi-arm polymeric conjugates of 7-ethyl-10-hydroxycamptothecin
CA2732508C (en) * 2008-08-11 2016-03-15 Nektar Therapeutics Multi-arm polymeric alkanoate conjugates
EP2501413B1 (en) * 2009-11-18 2019-03-27 Nektar Therapeutics Acid salt forms of polymer-drug conjugates
CN104784699B (en) * 2014-01-20 2019-05-03 博瑞生物医药(苏州)股份有限公司 Folate receptor binding ligand-drug conjugates
CN105396141B (en) * 2015-12-10 2018-08-24 浙江大学 IRGD- anticancer drug conjugates and its preparation method and application

Also Published As

Publication number Publication date
CN108727584A (en) 2018-11-02

Similar Documents

Publication Publication Date Title
AU2017265310B2 (en) Multi-arm polymeric targeting anti-cancer conjugate
AU2018255458B2 (en) Multi-arm targeted anti-cancer conjugate
CN108727583B (en) Multi-arm targeted anticancer conjugate
CN107854693B (en) Integrin receptor targeted anticancer conjugates
CN108727582B (en) Targeted anticancer conjugates
CN108727584B (en) Anti-cancer conjugates
CN109776787B (en) Multi-arm targeting conjugates
TWI771652B (en) CD44-targeted multi-arm conjugates
CN110152013B (en) Pectin-adriamycin conjugate and preparation method and application thereof
CN110418653B (en) Pectin-adriamycin conjugate and preparation method and application thereof
CN109771658B (en) Targeted multi-arm conjugates
CN109776788B (en) Folate receptor targeted multi-arm conjugates
WO2019096096A1 (en) Multi-arm targeting conjugate

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant